1
|
Guan Y, Ren Z, Yang B, Xu W, Wu W, Li X, Zhang T, Li D, Chen S, Bai J, Song X, Jia Z, Xiong X, He S, Li C, Meng F, Wu T, Zhang J, Liu X, Meng H, Peng J, Wang Y. Dual-bionic regenerative microenvironment for peripheral nerve repair. Bioact Mater 2023; 26:370-386. [PMID: 36942011 PMCID: PMC10024190 DOI: 10.1016/j.bioactmat.2023.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 03/18/2023] Open
Abstract
Autologous nerve grafting serves is considered the gold standard treatment for peripheral nerve defects; however, limited availability and donor area destruction restrict its widespread clinical application. Although the performance of allogeneic decellularized nerve implants has been explored, challenges such as insufficient human donors have been a major drawback to its clinical use. Tissue-engineered neural regeneration materials have been developed over the years, and researchers have explored strategies to mimic the peripheral neural microenvironment during the design of nerve catheter grafts, namely the extracellular matrix (ECM), which includes mechanical, physical, and biochemical signals that support nerve regeneration. In this study, polycaprolactone/silk fibroin (PCL/SF)-aligned electrospun material was modified with ECM derived from human umbilical cord mesenchymal stem cells (hUMSCs), and a dual-bionic nerve regeneration material was successfully fabricated. The results indicated that the developed biomimetic material had excellent biological properties, providing sufficient anchorage for Schwann cells and subsequent axon regeneration and angiogenesis processes. Moreover, the dual-bionic material exerted a similar effect to that of autologous nerve transplantation in bridging peripheral nerve defects in rats. In conclusion, this study provides a new concept for designing neural regeneration materials, and the prepared dual-bionic repair materials have excellent auxiliary regenerative ability and further preclinical testing is warranted to evaluate its clinical application potential.
Collapse
Affiliation(s)
- Yanjun Guan
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province, 226007, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Zhiqi Ren
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Boyao Yang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Wenjing Xu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Wenjun Wu
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Xiangling Li
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Tieyuan Zhang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Dongdong Li
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Shengfeng Chen
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jun Bai
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Xiangyu Song
- Hebei North University, Zhangjiakou, 075051, PR China
| | - Zhibo Jia
- Hebei North University, Zhangjiakou, 075051, PR China
| | - Xing Xiong
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Songlin He
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- School of Medicine, Nankai University, Tianjin, 300071, PR China
| | - Chaochao Li
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Fanqi Meng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Tong Wu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jian Zhang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Xiuzhi Liu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Haoye Meng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jiang Peng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province, 226007, PR China
- Corresponding author. Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China.
| | - Yu Wang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province, 226007, PR China
- Corresponding author. Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China.
| |
Collapse
|
2
|
Li Y, Zhang J, Chen L, Li H, Wang J. Repair of critical-sized rat cranial defects with RADA16-W9 self-assembled peptide hydrogel. Biochem Biophys Res Commun 2023; 652:68-75. [PMID: 36812709 DOI: 10.1016/j.bbrc.2023.02.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/26/2023] [Accepted: 02/12/2023] [Indexed: 02/15/2023]
Abstract
Bone defects are common in orthopaedics and there is an urgent need to explore effective bone repair materials with osteoinductive activity. Peptide self-assembled nanomaterials have a fibrous structure like that of the extracellular matrix and are ideal bionic scaffold materials. In this study, a short peptide WP9QY (W9) with strong osteoinductive effect was tagged to a self-assembled peptide RADA16 molecule through solid phase synthesis to design a RADA16-W9 peptide gel scaffold. A rat cranial defect was used as a research model to explore the effect of this peptide material on the repair of bone defects in vivo. The structure characteristic of the functional self-assembling peptide nanofiber hydrogel scaffold RADA16-W9 was evaluated by atomic force microscopy (AFM). Then adipose stem cells (ASCs) were isolated from Sprague-Dawley (SD) rat and cultured. the cellular compatibility of scaffold was evaluated through Live/Dead assay. Furthermore, we explore the effects of hydrogels in vivo with the critical-sized mouse calvarial defect model. Micro-CT analysis showed that the RADA16-W9 group had higher levels of bone volume/total volume (BV/TV) (P < 0.05),Trabecular number(TB.N) (P < 0.05),bone mineral density (BMD)(P < 0.05) and trabecular thickness (Tb. Th) (P < 0.05) compared with the RADA16 and PBS groups. Hematoxylin and eosin (H&E) staining showed that RADA16-W9 group had the highest bone regeneration level. Histochemical staining showed significantly higher expression levels of osteogenic factors such as alkaline phosphatase (ALP) and osteocalcin (OCN) in the RADA16-W9 group than in the other two groups (P < 0.05). Reverse transcription polymerase chain reaction (RT-PCR) quantification showed higher mRNA expression levels of osteogenic-related genes ALP, Runt-related transcription factor 2(Runx2), OCN, Osteopontin (OPN) in the RADA16-W9 group than in the RADA16 and PBS groups (P < 0.05). The live/dead staining results showed that RADA16-W9 is not toxic to rASCs and has good biocompatibility. In vivo experiments show that it accelerates the process of bone reconstruction, significantly promoting bone regeneration and can be used to develop a molecular drug for bone defect repair.
Collapse
Affiliation(s)
- Yong Li
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - JunKai Zhang
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - LiFu Chen
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - HaiTao Li
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Jian Wang
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
3
|
Griffanti G, McKee MD, Nazhat SN. Mineralization of Bone Extracellular Matrix-like Scaffolds Fabricated as Silk Sericin-Functionalized Dense Collagen–Fibrin Hybrid Hydrogels. Pharmaceutics 2023; 15:pharmaceutics15041087. [PMID: 37111573 PMCID: PMC10142947 DOI: 10.3390/pharmaceutics15041087] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
The design of hydrogels that combine both the biochemical cues needed to direct seeded cellular functions and mineralization to provide the structural and mechanical properties approaching those of mineralized native bone extracellular matrix (ECM) represents a significant challenge in bone tissue engineering. While fibrous hydrogels constituting of collagen or fibrin (and their hybrids) can be considered as scaffolds that mimic to some degree native bone ECM, their insufficient mechanical properties limit their application. In the present study, an automated gel aspiration–ejection (automated GAE) method was used to generate collagen–fibrin hybrid gel scaffolds with micro-architectures and mechanical properties approaching those of native bone ECM. Moreover, the functionalization of these hybrid scaffolds with negatively charged silk sericin accelerated their mineralization under acellular conditions in simulated body fluid and modulated the proliferation and osteoblastic differentiation of seeded MC3T3-E1 pre-osteoblastic cells. In the latter case, alkaline phosphatase activity measurements indicated that the hybrid gel scaffolds with seeded cells showed accelerated osteoblastic differentiation, which in turn led to increased matrix mineralization. In summary, the design of dense collagen–fibrin hybrid gels through an automated GAE process can provide a route to tailoring specific biochemical and mechanical properties to different types of bone ECM-like scaffolds, and can provide a model to better understand cell–matrix interactions in vitro for bioengineering purposes.
Collapse
Affiliation(s)
- Gabriele Griffanti
- Department of Mining and Materials Engineering, McGill University, Montréal, QC H3A 0C5, Canada;
| | - Marc D. McKee
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada;
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada
| | - Showan N. Nazhat
- Department of Mining and Materials Engineering, McGill University, Montréal, QC H3A 0C5, Canada;
- Correspondence: ; Tel.: +514-398-5524; Fax: 514-398-4492
| |
Collapse
|
4
|
Zhao H, Liu C, Liu Y, Ding Q, Wang T, Li H, Wu H, Ma T. Harnessing electromagnetic fields to assist bone tissue engineering. Stem Cell Res Ther 2023; 14:7. [PMID: 36631880 PMCID: PMC9835389 DOI: 10.1186/s13287-022-03217-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/08/2022] [Indexed: 01/13/2023] Open
Abstract
Bone tissue engineering (BTE) emerged as one of the exceptional means for bone defects owing to it providing mechanical supports to guide bone tissue regeneration. Great advances have been made to facilitate the success of BTE in regenerating bone within defects. The use of externally applied fields has been regarded as an alternative strategy for BTE. Electromagnetic fields (EMFs), known as a simple and non-invasive therapy, can remotely provide electric and magnetic stimulation to cells and biomaterials, thus applying EMFs to assist BTE would be a promising strategy for bone regeneration. When combined with BTE, EMFs improve cell adhesion to the material surface by promoting protein adsorption. Additionally, EMFs have positive effects on mesenchymal stem cells and show capabilities of pro-angiogenesis and macrophage polarization manipulation. These advantages of EMFs indicate that it is perfectly suitable for representing the adjuvant treatment of BTE. We also summarize studies concerning combinations of EMFs and diverse biomaterial types. The strategy of combining EMFs and BTE receives encouraging outcomes and holds a promising future for effectively treating bone defects.
Collapse
Affiliation(s)
- Hongqi Zhao
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Chaoxu Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Yang Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Qing Ding
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Tianqi Wang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Hao Li
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
5
|
Sefkow-Werner J, Le Pennec J, Machillot P, Ndayishimiye B, Castro-Ramirez E, Lopes J, Licitra C, Wang I, Delon A, Picart C, Migliorini E. Automated Fabrication of Streptavidin-Based Self-assembled Materials for High-Content Analysis of Cellular Response to Growth Factors. ACS APPLIED MATERIALS & INTERFACES 2022; 14:10.1021/acsami.2c08272. [PMID: 35849638 PMCID: PMC7614070 DOI: 10.1021/acsami.2c08272] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The automation of liquid-handling routines offers great potential for fast, reproducible, and labor-reduced biomaterial fabrication but also requires the development of special protocols. Competitive systems demand for a high degree in miniaturization and parallelization while maintaining flexibility regarding the experimental design. Today, there are only a few possibilities for automated fabrication of biomaterials inside multiwell plates. We have previously demonstrated that streptavidin-based biomimetic platforms can be employed to study cellular behaviors on biomimetic surfaces. So far, these self-assembled materials were made by stepwise assembly of the components using manual pipetting. In this work, we introduce for the first time a fully automated and adaptable workflow to functionalize glass-bottom multiwell plates with customized biomimetic platforms deposited in single wells using a liquid-handling robot. We then characterize the cell response using automated image acquisition and subsequent analysis. Furthermore, the molecular surface density of the biomimetic platforms was characterized in situ using fluorescence-based image correlation spectroscopy. These measurements were in agreement with standard ex situ spectroscopic ellipsometry measurements. Due to automation, we could do a proof of concept to study the effect of heparan sulfate on the bioactivity of bone morphogenetic proteins on myoblast cells, using four different bone morphogenetic proteins (BMPs) (2, 4, 6, and 7) in parallel, at five increasing concentrations. Using such an automated self-assembly of biomimetic materials, it may be envisioned to further investigate the role of a large variety of extracellular matrix (ECM) components and growth factors on cell signaling.
Collapse
Affiliation(s)
- Julius Sefkow-Werner
- Univ. Grenoble Alpes, CNRS, Grenoble INP**, LMGP, 38000 Grenoble, France
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Jean Le Pennec
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Paul Machillot
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Bertin Ndayishimiye
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Elaine Castro-Ramirez
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Joao Lopes
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | | | - Irene Wang
- Univ. Grenoble Alpes, CNRS, LiPhy, Grenoble, France
| | | | - Catherine Picart
- Univ. Grenoble Alpes, CNRS, Grenoble INP**, LMGP, 38000 Grenoble, France
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| | - Elisa Migliorini
- Univ. Grenoble Alpes, CNRS, Grenoble INP**, LMGP, 38000 Grenoble, France
- Univ. Grenoble Alpes, CEA, INSERM, U1292 Biosanté, CNRS EMR 5000 BRM, 3800, Grenoble, France
| |
Collapse
|
6
|
Zha K, Tian Y, Panayi AC, Mi B, Liu G. Recent Advances in Enhancement Strategies for Osteogenic Differentiation of Mesenchymal Stem Cells in Bone Tissue Engineering. Front Cell Dev Biol 2022; 10:824812. [PMID: 35281084 PMCID: PMC8904963 DOI: 10.3389/fcell.2022.824812] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Although bone is an organ that displays potential for self-healing after damage, bone regeneration does not occur properly in some cases, and it is still a challenge to treat large bone defects. The development of bone tissue engineering provides a new approach to the treatment of bone defects. Among various cell types, mesenchymal stem cells (MSCs) represent one of the most promising seed cells in bone tissue engineering due to their functions of osteogenic differentiation, immunomodulation, and secretion of cytokines. Regulation of osteogenic differentiation of MSCs has become an area of extensive research over the past few years. This review provides an overview of recent research progress on enhancement strategies for MSC osteogenesis, including improvement in methods of cell origin selection, culture conditions, biophysical stimulation, crosstalk with macrophages and endothelial cells, and scaffolds. This is favorable for further understanding MSC osteogenesis and the development of MSC-based bone tissue engineering.
Collapse
Affiliation(s)
- Kangkang Zha
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yue Tian
- Department of Military Patient Management, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Institute of Orthopaedics, Chinese PLA General Hospital, Beijing, China
| | - Adriana C. Panayi
- Division of Plastic Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
- *Correspondence: Bobin Mi, ; Guohui Liu,
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
- *Correspondence: Bobin Mi, ; Guohui Liu,
| |
Collapse
|
7
|
Kurian AG, Singh RK, Patel KD, Lee JH, Kim HW. Multifunctional GelMA platforms with nanomaterials for advanced tissue therapeutics. Bioact Mater 2022; 8:267-295. [PMID: 34541401 PMCID: PMC8424393 DOI: 10.1016/j.bioactmat.2021.06.027] [Citation(s) in RCA: 155] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Polymeric hydrogels are fascinating platforms as 3D scaffolds for tissue repair and delivery systems of therapeutic molecules and cells. Among others, methacrylated gelatin (GelMA) has become a representative hydrogel formulation, finding various biomedical applications. Recent efforts on GelMA-based hydrogels have been devoted to combining them with bioactive and functional nanomaterials, aiming to provide enhanced physicochemical and biological properties to GelMA. The benefits of this approach are multiple: i) reinforcing mechanical properties, ii) modulating viscoelastic property to allow 3D printability of bio-inks, iii) rendering electrical/magnetic property to produce electro-/magneto-active hydrogels for the repair of specific tissues (e.g., muscle, nerve), iv) providing stimuli-responsiveness to actively deliver therapeutic molecules, and v) endowing therapeutic capacity in tissue repair process (e.g., antioxidant effects). The nanomaterial-combined GelMA systems have shown significantly enhanced and extraordinary behaviors in various tissues (bone, skin, cardiac, and nerve) that are rarely observable with GelMA. Here we systematically review these recent efforts in nanomaterials-combined GelMA hydrogels that are considered as next-generation multifunctional platforms for tissue therapeutics. The approaches used in GelMA can also apply to other existing polymeric hydrogel systems.
Collapse
Affiliation(s)
- Amal George Kurian
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Rajendra K. Singh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Kapil D. Patel
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, London, WC1X8LD, UK
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| |
Collapse
|
8
|
Petre DG, Leeuwenburgh SCG. The Use of Fibers in Bone Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:141-159. [PMID: 33375900 DOI: 10.1089/ten.teb.2020.0252] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Bone tissue engineering aims to restore and maintain the function of bone by means of biomaterial-based scaffolds. This review specifically focuses on the use of fibers in biomaterials used for bone tissue engineering as suitable environment for bone tissue repair and regeneration. We present a bioinspired rationale behind the use of fibers in bone tissue engineering and provide an overview of the most common fiber fabrication methods, including solution, melt, and microfluidic spinning. Subsequently, we provide a brief overview of the composition of fibers that are used in bone tissue engineering, including fibers composed of (i) natural polymers (e.g., cellulose, collagen, gelatin, alginate, chitosan, and silk, (ii) synthetic polymers (e.g., polylactic acid [PLA], polycaprolactone, polyglycolic acid [PGA], polyethylene glycol, and polymer blends of PLA and PGA), (iii) ceramic fibers (e.g., aluminium oxide, titanium oxide, and zinc oxide), (iv) metallic fibers (e.g., titanium and its alloys, copper and magnesium), and (v) composite fibers. In addition, we review the most relevant fiber modification strategies that are used to enhance the (bio)functionality of these fibers. Finally, we provide an overview of the applicability of fibers in biomaterials for bone tissue engineering, with a specific focus on mechanical, pharmaceutical, and biological properties of fiber-functionalized biomaterials for bone tissue engineering. Impact statement Natural bone is a complex composite material composed of an extracellular matrix of mineralized fibers containing living cells and bioactive molecules. Consequently, the use of fibers in biomaterial-based scaffolds offers a wide variety of opportunities to replicate the functional performance of bone. This review provides an overview of the use of fibers in biomaterials for bone tissue engineering, thereby contributing to the design of novel fiber-functionalized bone-substituting biomaterials of improved functionality regarding their mechanical, pharmaceutical, and biological properties.
Collapse
Affiliation(s)
- Daniela Geta Petre
- Department of Dentistry-Regenerative Biomaterials, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Sander C G Leeuwenburgh
- Department of Dentistry-Regenerative Biomaterials, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
9
|
Evdokimov PV, Tikhonova SA, Kiseleva AK, Filippov YY, Novoseletskaya ES, Efimenko AY, Putlayev VI. Effect of the Pore Size on the Biological Activity of β-Ca3(PO4)2-Based Resorbable Macroporous Ceramic Materials Obtained by Photopolymerization. RUSS J INORG CHEM+ 2021. [PMCID: PMC8601372 DOI: 10.1134/s0036023621110061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract
The effect of the pore size of macroporous ceramic materials based on β-Ca3(PO4)2 on their biological activity was studied. The formation conditions of macroporous ceramics with a porosity of >50% and a specified pore size were determined. The effect of components of the light-curing emulsion on the pore size in the final macroporous ceramics was studied. The biocompatibility of β-Ca3(PO4)2-based macroporous ceramics was demonstrated in in vitro biomedical assays. The effect of pore size of macroporous ceramic materials on mesenchymal stromal cell proliferation and viability was established.
Collapse
Affiliation(s)
- P. V. Evdokimov
- Kurnakov Institute of General and Inorganic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russia
- Moscow State University, 119991 Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
10
|
Wu M, Zou L, Jiang L, Zhao Z, Liu J. Osteoinductive and antimicrobial mechanisms of graphene-based materials for enhancing bone tissue engineering. J Tissue Eng Regen Med 2021; 15:915-935. [PMID: 34469046 DOI: 10.1002/term.3239] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 02/05/2023]
Abstract
Graphene-based materials (GMs) have great application prospects in bone tissue engineering due to their osteoinductive ability and antimicrobial activity. GMs induce osteogenic differentiation through several mechanisms and pathways in bone tissue engineering. First of all, the surface and high hardness of the porous folds of graphene or graphene oxide (GO) can generate mechanical stimulation to initiate a cascade of reactions that promote osteogenic differentiation without any chemical inducers. In addition, change of the extracellular matrix (ECM), regulation of macrophage polarization, the oncostatin M (OSM) signaling pathway, the MAPK signaling pathway, the BMP signaling pathway, the Wnt/β-catenin signaling pathway, and other pathways are involved in GMs' regulation of osteogenesis. In bone tissue engineering, GMs prevent the formation of microbial biofilms mainly through preventing microbial adhesion and killing them. The former is mainly achieved by reducing surface free energy (SFE) and increasing hydrophobicity. The latter mainly includes oxidative stress and photothermal/photodynamic effects. Graphene and its derivatives (GDs) are mainly combined with bioactive ceramic materials, metal materials and macromolecular polymers to play an antimicrobial effect in bone tissue engineering. Concentration, number of layers, and type of GDs often affect the antimicrobial activity of GMs. In this paper, we reviewed relevant osteoinductive and antimicrobial mechanisms of GMs and their applications in bone tissue engineering.
Collapse
Affiliation(s)
- Mengsong Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linli Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Fracture Healing Research-Shift towards In Vitro Modeling? Biomedicines 2021; 9:biomedicines9070748. [PMID: 34203470 PMCID: PMC8301383 DOI: 10.3390/biomedicines9070748] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 01/07/2023] Open
Abstract
Fractures are one of the most frequently occurring traumatic events worldwide. Approximately 10% of fractures lead to bone healing disorders, resulting in strain for affected patients and enormous costs for society. In order to shed light into underlying mechanisms of bone regeneration (habitual or disturbed), and to develop new therapeutic strategies, various in vivo, ex vivo and in vitro models can be applied. Undeniably, in vivo models include the systemic and biological situation. However, transferability towards the human patient along with ethical concerns regarding in vivo models have to be considered. Fostered by enormous technical improvements, such as bioreactors, on-a-chip-technologies and bone tissue engineering, sophisticated in vitro models are of rising interest. These models offer the possibility to use human cells from individual donors, complex cell systems and 3D models, therefore bridging the transferability gap, providing a platform for the introduction of personalized precision medicine and finally sparing animals. Facing diverse processes during fracture healing and thus various scientific opportunities, the reliability of results oftentimes depends on the choice of an appropriate model. Hence, we here focus on categorizing available models with respect to the requirements of the scientific approach.
Collapse
|
12
|
Du T, Niu X, Hou S, Xu M, Li Z, Li P, Fan Y. Highly aligned hierarchical intrafibrillar mineralization of collagen induced by periodic fluid shear stress. J Mater Chem B 2021; 8:2562-2572. [PMID: 32101230 DOI: 10.1039/c9tb02643f] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Periodic fluid shear stress (FSS) is one of the main mechanical microenvironments in mineralization of bone matrix. To elucidate the mechanism of periodic FSS in collagen mineralization, a mechanical loading induced mineralization system is developed and compared with traditional polyacrylic acid (PAA) induced mineralization. Fourier transform infrared (FTIR) spectroscopy, calcium-to-phosphorus molar ratio and transmission electron microscopy (TEM) demonstrate that both periodic FSS and PAA can control the size of amorphous calcium phosphate (ACP) to avoid aggregation and help the formation of intrafibrillar mineralization. Differently, periodic FSS under a proper cycle and range can accelerate the conversion of ACP to apatite crystals and alleviate the reduced transformation caused by PAA. Under the action of template analogues, periodic FSS can also promote the formation of highly oriented hierarchical intrafibrillar mineralized (HIM) collagen. These findings are helpful for understanding the mechanism of collagen mineralization in natural bone matrix and contribute to the design of novel bone substitute materials with hierarchical structures.
Collapse
Affiliation(s)
- Tianming Du
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Xufeng Niu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China. and Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China and Research Institute of Beihang University in Shenzhen, Shenzhen 518057, China
| | - Sen Hou
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China. and Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
| | - Menghan Xu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Zhengwei Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | - Ping Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China. and Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China. and Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing 100083, China and Beijing Key Laboratory of Rehabilitation Technical Aids for Old-Age Disability, National Research Center for Rehabilitation Technical Aids, Beijing 100176, China
| |
Collapse
|
13
|
Antipova CG, Lukanina KI, Krasheninnikov SV, Malakhov SN, Kamyshinsky RA, Grigoriev TE, Chvalun SN. Study of highly porous poly‐
l
‐lactide‐based composites with chitosan and collagen. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Christina G. Antipova
- Department of Nanobiomaterials and Structures National Research Centre “Kurchatov Institute” Moscow Russia
| | - Ksenia I. Lukanina
- Department of Nanobiomaterials and Structures National Research Centre “Kurchatov Institute” Moscow Russia
| | - Sergey V. Krasheninnikov
- Department of Nanobiomaterials and Structures National Research Centre “Kurchatov Institute” Moscow Russia
| | - Sergey N. Malakhov
- Department for Resource Centre National Research Centre “Kurchatov Institute” Moscow Russia
| | - Roman A. Kamyshinsky
- Resource Centre for Probe and Electron Microscopy National Research Centre “Kurchatov Institute” Moscow Russia
- Shubnikov Institute of Crystallography of Federal Scientific Research Centre “Crystallography and Photonics” Russian Academy of Sciences Moscow Russia
| | - Timofei E. Grigoriev
- Department of Nanobiomaterials and Structures National Research Centre “Kurchatov Institute” Moscow Russia
| | - Sergey N. Chvalun
- Department of Nanobiomaterials and Structures National Research Centre “Kurchatov Institute” Moscow Russia
| |
Collapse
|
14
|
Gavazzo P, Viti F, Donnelly H, Oliva MAG, Salmeron-Sanchez M, Dalby MJ, Vassalli M. Biophysical phenotyping of mesenchymal stem cells along the osteogenic differentiation pathway. Cell Biol Toxicol 2021; 37:915-933. [PMID: 33420657 DOI: 10.1007/s10565-020-09569-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/30/2020] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cells represent an important resource, for bone regenerative medicine and therapeutic applications. This review focuses on new advancements and biophysical tools which exploit different physical and chemical markers of mesenchymal stem cell populations, to finely characterize phenotype changes along their osteogenic differentiation process. Special attention is paid to recently developed label-free methods, which allow monitoring cell populations with minimal invasiveness. Among them, quantitative phase imaging, suitable for single-cell morphometric analysis, and nanoindentation, functional to cellular biomechanics investigation. Moreover, the pool of ion channels expressed in cells during differentiation is discussed, with particular interest for calcium homoeostasis.Altogether, a biophysical perspective of osteogenesis is proposed, offering a valuable tool for the assessment of the cell stage, but also suggesting potential physiological links between apparently independent phenomena.
Collapse
Affiliation(s)
- Paola Gavazzo
- Institute of Biophysics, National Research Council, Genoa, Italy
| | - Federica Viti
- Institute of Biophysics, National Research Council, Genoa, Italy.
| | - Hannah Donnelly
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Mariana Azevedo Gonzalez Oliva
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, UK
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Massimo Vassalli
- Centre for the Cellular Microenvironment, Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, UK
| |
Collapse
|
15
|
Chen G, Kong P, Jiang A, Wang X, Sun Y, Yu T, Chi H, Song C, Zhang H, Subedi D, Ravi Kumar P, Bai K, Liu K, Ji Y, Yan J. A modular programmed biphasic dual-delivery system on 3D ceramic scaffolds for osteogenesis in vitro and in vivo. J Mater Chem B 2020; 8:9697-9717. [PMID: 32789334 DOI: 10.1039/c9tb02127b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Single-factor delivery is the most common characteristic of bone tissue engineering techniques. However, bone regeneration is a complex process requiring multiple factors and specialized release mechanisms. Therefore, the development of a dual-delivery system allowing for programmed release kinetics would be highly desirable. Improvement of the molarity and versatility of the delivery system has rarely been studied. Herein, we report the development of a novel, modular programmed biphasic dual-release system (SCB), carrying a BMP2 and an engineered collagen I-derived recognition motif (Stath-DGEA), with a self-remodification feature on hydroxyapatite (HA)-based materials. The SCB system was loaded onto an additive manufactured (AM) scaffold in order to evaluate its bifactor osteogenic potential and its biphasic release behavior. Further, the biomechanical properties of the scaffold were studied by using the fluid-structure interaction (FSI) method. Section fluorescent labeling revealed that the HA scaffold has a relatively higher density and efficiency. Additionally, the results of the release and inhibition experiment suggested that the SCB system could facilitate the sustained release of therapeutic levels of two factors during the initial stage of implantation, thereby exhibiting a rapid high-dose release pattern at a specific time point during the second stage. The FSI prediction model indicated that the scaffold provides an excellent biomimetic mechanical and fluid dynamic microenvironment to promote osteogenesis. Our results indicated that incorporation of BMP2 with Stath-DGEA in the biphasic SCB system could have a synergetic effect in promoting the adhesion, proliferation, and differentiation of bone marrow mesenchymal stem cells (BMSCs) in vitro, under staged stimulations. Further, in vivo studies in both ectopic and orthotopic rat models showed that the SCB system loaded onto an AM scaffold could enhance osteointegration and osteoinduction throughout the osteogenic process. Thus, the novel synthetic SCB system described herein used on an AM scaffold provides a biomimetic extracellular environment that enhances bone regeneration and is a promising multifunctional, dual-release platform.
Collapse
Affiliation(s)
- Guanghua Chen
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Fischer NG, Münchow EA, Tamerler C, Bottino MC, Aparicio C. Harnessing biomolecules for bioinspired dental biomaterials. J Mater Chem B 2020; 8:8713-8747. [PMID: 32747882 PMCID: PMC7544669 DOI: 10.1039/d0tb01456g] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dental clinicians have relied for centuries on traditional dental materials (polymers, ceramics, metals, and composites) to restore oral health and function to patients. Clinical outcomes for many crucial dental therapies remain poor despite many decades of intense research on these materials. Recent attention has been paid to biomolecules as a chassis for engineered preventive, restorative, and regenerative approaches in dentistry. Indeed, biomolecules represent a uniquely versatile and precise tool to enable the design and development of bioinspired multifunctional dental materials to spur advancements in dentistry. In this review, we survey the range of biomolecules that have been used across dental biomaterials. Our particular focus is on the key biological activity imparted by each biomolecule toward prevention of dental and oral diseases as well as restoration of oral health. Additional emphasis is placed on the structure-function relationships between biomolecules and their biological activity, the unique challenges of each clinical condition, limitations of conventional therapies, and the advantages of each class of biomolecule for said challenge. Biomaterials for bone regeneration are not reviewed as numerous existing reviews on the topic have been recently published. We conclude our narrative review with an outlook on the future of biomolecules in dental biomaterials and potential avenues of innovation for biomaterial-based patient oral care.
Collapse
Affiliation(s)
- Nicholas G Fischer
- Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, 16-250A Moos Tower, 515 Delaware St. SE, Minneapolis, Minnesota 55455, USA.
| | | | | | | | | |
Collapse
|
17
|
Chang PC, Luo HT, Lin ZJ, Tai WC, Chang CH, Chang YC, Cochran DL, Chen MH. Regeneration of critical-sized mandibular defect using a 3D-printed hydroxyapatite-based scaffold: An exploratory study. J Periodontol 2020; 92:428-435. [PMID: 32761906 DOI: 10.1002/jper.20-0110] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/19/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Three-dimensional (3D) printing has become an available technology to fabricate customized tissue engineering scaffolds with delicate architecture. This exploratory study aimed to evaluate the potential of a 3D-printed hydroxyapatite-based scaffold as a biomaterial for obtaining guided bone regeneration (GBR) in vivo. METHODS Scaffolds composed of 90% hydroxyapatite and 10% poly(lactic-co-glycolic acid) were printed using a microextrusion process to fit 4 mm diameter and 0.5 mm thick through-and-through osseous defects on the mandibular ramus of rats, with unfilled defects serving as controls. Specimens were analyzed for regeneration-associated gene expression on day 7, and micro-computed tomography (micro-CT) and histology assessments were carried out on day 28. RESULTS The scaffolds were 3.56 ± 0.43 mm (x-axis) and 4.02 ± 0.44 mm (y-axis) in diameter and 0.542 ± 0.035 mm thick (z-axis), with a mean pore size of 0.420 ± 0.028 × 0.328 ± 0.005 mm2 . Most scaffolds fit the defects well. Type I collagen, VEGF, and Cbfa1 were upregulated in the scaffold-treated defects by day 7. By day 28, de novo osteogenesis and scaffold-tissue integration were evident in the scaffold-treated defects, and entire mineralized tissue, as well as newly formed bone, was significantly promoted, as seen in the micro-CT and histologic analyses. CONCLUSION The 3D-printed hydroxyapatite-based scaffold showed acceptable dimensional stability and demonstrated favorable osteoregenerative capability that fulfilled the need for GBR.
Collapse
Affiliation(s)
- Po-Chun Chang
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.,Division of Periodontics, Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan.,School of Denstistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Ting Luo
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan.,Division of Periodontics, Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Zhi-Jie Lin
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Wei-Chiu Tai
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Ching-He Chang
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Ying-Chieh Chang
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - David L Cochran
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Min-Huey Chen
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
18
|
Sefkow-Werner J, Machillot P, Sales A, Castro-Ramirez E, Degardin M, Boturyn D, Cavalcanti-Adam EA, Albiges-Rizo C, Picart C, Migliorini E. Heparan sulfate co-immobilized with cRGD ligands and BMP2 on biomimetic platforms promotes BMP2-mediated osteogenic differentiation. Acta Biomater 2020; 114:90-103. [PMID: 32673751 DOI: 10.1016/j.actbio.2020.07.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/27/2022]
Abstract
The chemical and physical properties of the extracellular matrix (ECM) are known to be fundamental for regulating growth factor bioactivity. The role of heparan sulfate (HS), a glycosaminoglycan, and of cell adhesion proteins (containing the cyclic RGD (cRGD) ligands) on bone morphogenetic protein 2 (BMP2)-mediated osteogenic differentiation has not been fully explored. In particular, it is not known whether and how their effects can be potentiated when they are presented in controlled close proximity, as in the ECM. Here, we developed streptavidin platforms to mimic selective aspects of the in vivo presentation of cRGD, HS and BMP2, with a nanoscale-control of their surface density and orientation to study cell adhesion and osteogenic differentiation. We showed that whereas a controlled increase in cRGD surface concentration upregulated BMP2 signaling due to β3 integrin recruitment, silencing either β1 or β3 integrins negatively affected BMP2-mediated phosphorylation of SMAD1/5/9 and alkaline phosphatase expression. Furthermore, the presence of adsorbed BMP2 promoted cellular adhesion at very low cRGD concentrations. Finally, we proved that HS co-immobilized with cRGD both sustained BMP2 signaling and enhanced osteogenic differentiation compared to BMP2 directly immobilized on streptavidin, even with a low cRGD surface concentration. Altogether, our results show that HS facilitated and sustained the synergy between BMP2 and integrin pathways and that the co-immobilization of HS and cRGD peptides optimised BMP2-mediated osteogenic differentiation. Statement of significance The growth factor BMP2 is used to treat large bone defects. Previous studies have shown that the presentation of BMP2 via extracellular matrix molecules, such as heparan sulfate (HS), can upregulate BMP2 signaling. The potential advantages of dose reduction and local specificity have stimulated interest in further investigations into biomimetic approaches. We designed a streptavidin model surface eligible for immobilizing tunable amounts of molecules from the extracellular space, such as HS, adhesion motifs (cyclic RGD) and BMP2. By studying cellular adhesion, BMP2 bioactivity and its osteogenic potential we reveal the combined effect of integrins, HS and BMP2, which contribute in answering fundamental questions regarding cell-matrix interaction.
Collapse
|
19
|
Trujillo S, Gonzalez-Garcia C, Rico P, Reid A, Windmill J, Dalby MJ, Salmeron-Sanchez M. Engineered 3D hydrogels with full-length fibronectin that sequester and present growth factors. Biomaterials 2020; 252:120104. [DOI: 10.1016/j.biomaterials.2020.120104] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/22/2022]
|
20
|
Borciani G, Montalbano G, Baldini N, Cerqueni G, Vitale-Brovarone C, Ciapetti G. Co-culture systems of osteoblasts and osteoclasts: Simulating in vitro bone remodeling in regenerative approaches. Acta Biomater 2020; 108:22-45. [PMID: 32251782 DOI: 10.1016/j.actbio.2020.03.043] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 02/08/2023]
Abstract
Bone is an extremely dynamic tissue, undergoing continuous remodeling for its whole lifetime, but its regeneration or augmentation due to bone loss or defects are not always easy to obtain. Bone tissue engineering (BTE) is a promising approach, and its success often relies on a "smart" scaffold, as a support to host and guide bone formation through bone cell precursors. Bone homeostasis is maintained by osteoblasts (OBs) and osteoclasts (OCs) within the basic multicellular unit, in a consecutive cycle of resorption and formation. Therefore, a functional scaffold should allow the best possible OB/OC cooperation for bone remodeling, as happens within the bone extracellular matrix in the body. In the present work OB/OC co-culture models, with and without scaffolds, are reviewed. These experimental systems are intended for different targets, including bone remodeling simulation, drug testing and the assessment of biomaterials and 3D scaffolds for BTE. As a consequence, several parameters, such as cell type, cell ratio, culture medium and inducers, culture times and setpoints, assay methods, etc. vary greatly. This review identifies and systematically reports the in vitro methods explored up to now, which, as they allow cellular communication, more closely resemble bone remodeling and/or the regeneration process in the framework of BTE. STATEMENT OF SIGNIFICANCE: Bone is a dynamic tissue under continuous remodeling, but spontaneous healing may fail in the case of excessive bone loss which often requires valid alternatives to conventional treatments to restore bone integrity, like bone tissue engineering (BTE). Pre-clinical evaluation of scaffolds for BTE requires in vitro testing where co-cultures combining innovative materials with osteoblasts (OBs) and osteoclasts (OCs) closely mimic the in vivo repair process. This review considers the direct and indirect OB/OC co-cultures relevant to BTE, from the early mouse-cell models to the recent bone regenerative systems. The co-culture modeling of bone microenvironment provides reliable information on bone cell cross-talk. Starting from improved knowledge on bone remodeling, bone disease mechanisms may be understood and new BTE solutions are designed.
Collapse
|
21
|
Newby SD, Masi T, Griffin CD, King WJ, Chipman A, Stephenson S, Anderson DE, Biris AS, Bourdo SE, Dhar M. Functionalized Graphene Nanoparticles Induce Human Mesenchymal Stem Cells to Express Distinct Extracellular Matrix Proteins Mediating Osteogenesis. Int J Nanomedicine 2020; 15:2501-2513. [PMID: 32368037 PMCID: PMC7171876 DOI: 10.2147/ijn.s245801] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The extracellular matrix (ECM) labyrinthine network secreted by mesenchymal stem cells (MSCs) provides a microenvironment that enhances cell adherence, proliferation, viability, and differentiation. The potential of graphene-based nanomaterials to mimic a tissue-specific ECM has been recognized in designing bone tissue engineering scaffolds. In this study, we investigated the expression of specific ECM proteins when human fat-derived adult MSCs adhered and underwent osteogenic differentiation in the presence of functionalized graphene nanoparticles. METHODS Graphene nanoparticles with 6-10% oxygen content were prepared and characterized by XPS, FTIR, AFM and Raman spectroscopy. Calcein-am and crystal violet staining were performed to evaluate viability and proliferation of human fat-derived MSCs on graphene nanoparticles. Alizarin red staining and quantitation were used to determine the effect of graphene nanoparticles on osteogenic differentiation. Finally, immunofluorescence assays were used to investigate the expression of ECM proteins during cell adhesion and osteogenic differentiation. RESULTS Our data show that in the presence of graphene, MSCs express specific integrin heterodimers and exhibit a distinct pattern of the corresponding bone-specific ECM proteins, primarily fibronectin, collagen I and vitronectin. Furthermore, MSCs undergo osteogenic differentiation spontaneously without any chemical induction, suggesting that the physicochemical properties of graphene nanoparticles might trigger the expression of bone-specific ECM. CONCLUSION Understanding the cell-graphene interactions resulting in an osteogenic niche for MSCs will significantly improve the application of graphene nanoparticles in bone repair and regeneration.
Collapse
Affiliation(s)
- Steven D Newby
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN37996, USA
- Comparative and Experimental Medicine, University of Tennessee, Knoxville, TN37996, USA
| | - Tom Masi
- University of Tennessee Graduate School of Medicine, Knoxville, TN37996, USA
| | - Christopher D Griffin
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR72204, USA
| | - William J King
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR72204, USA
| | - Anna Chipman
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN37996, USA
| | - Stacy Stephenson
- University of Tennessee Graduate School of Medicine, Knoxville, TN37996, USA
| | - David E Anderson
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN37996, USA
| | - Alexandru S Biris
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR72204, USA
| | - Shawn E Bourdo
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, Little Rock, AR72204, USA
| | - Madhu Dhar
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN37996, USA
| |
Collapse
|
22
|
Liu Y, Yang S, Cao L, Zhang X, Wang J, Liu C. Facilitated vascularization and enhanced bone regeneration by manipulation hierarchical pore structure of scaffolds. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110622. [PMID: 32204064 DOI: 10.1016/j.msec.2019.110622] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 12/29/2019] [Accepted: 12/31/2019] [Indexed: 12/31/2022]
Abstract
Sufficient vascularization is quite important for preventing cell death and promoting host integration during the repair of the critical sized bone defects. Porous structure providing enough space for the ingrowth of vessels is an essential consideration during the scaffold's development. In this study, we designed and fabricated three kinds of porous structured scaffolds based on poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) (PHBHHx), such as mono-structured PHBHHx scaffolds with macro pores (PH-1), di-structured PHBHHx scaffolds with macro-meso pores (PHS-2), and tri-structured PHBHHx scaffolds with macro-micro-meso pores (PHS-3), respectively. In vitro effects of the hierarchical porous scaffolds on human umbilical vein endothelial cells (HUVECs), such as cell attachment, glucose and lactate detection, relative gene expressions of endothelial markers were investigated. The PHS-3 scaffolds exhibited preferential potency of inducing better angiogenesis in vitro. Consequently, the hierarchical porous scaffolds were applied to load rhBMP-2 and repair the critical sized bone defect (15 mm) in rabbits. Microangiography analysis by three dimensional micro-computed tomographic (micro-CT) demonstrated that the volume of blood vessels within the defect area was higher in the rhBMP-2 loaded PHS-3 (PHS-3/rhBMP-2) than that in other rhBMP-2 loaded porous scaffolds with simplex or double scaled pores (PH-1/rhBMP-2 or PHS-2/rhBMP-2) at 4 weeks and 8 weeks, which implied that multi-level porous structure was conducive to nutrition transmission and revascularization. Further investigations of orthotopic bone formation by micro-CT, histological and immunohistochemistry analysis confirmed the most accelerated new bone formation rate in the PHS-3/rhBMP-2 group. The maximum load value of the regenerated bone induced by PHS-3/rhBMP-2 at 12 weeks was 258.47 ± 14.77 N which did not show significant difference from the normal bone of 268.81 ± 12.05 N. These results highlighted that introducing multi-level pores into the biocompatible scaffolds may be an effective approach to promote angiogenesis and bone regeneration.
Collapse
Affiliation(s)
- Yang Liu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Shengbing Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, People's Republic of China
| | - Lingyan Cao
- Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200011, People's Republic of China
| | - Xiaohui Zhang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, People's Republic of China; Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China.
| | - Changsheng Liu
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China; Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China.
| |
Collapse
|
23
|
Zennifer A, Sekar MP, Subramanian A, Sethuraman S. Nanofiber matrices of protein mimetic bioactive peptides for biomedical applications. ARTIFICIAL PROTEIN AND PEPTIDE NANOFIBERS 2020:199-217. [DOI: 10.1016/b978-0-08-102850-6.00009-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
24
|
Augmented osteogenesis of mesenchymal stem cells using a fragmented Runx2 mixed with cell-penetrating, dimeric a-helical peptide. Eur J Pharm Sci 2019; 144:105210. [PMID: 31899341 DOI: 10.1016/j.ejps.2019.105210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/15/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022]
Abstract
The intracellular delivery of transcription factor/cofactor using cell penetrating peptide (CPP) can lead to selective osteogenesis. The present work investigates the cell-penetrating potential of the a cyclic, α-helical cell-penetrating peptide based on leucine and lysine residues (cLK) for intracellular delivery in MC3T3 cells and the osteogenic effects of a C-terminal proline‑serine‑threonine-rich (PST) domain of Runx2 using cLK in rat mesenchymal stem cells (MSCs). We confirmed that the combination of cLK and fluorescein 5-isothiocyanate (FITC)-fragmented-Runx2 (fRunx2) showed an enhanced cell-penetrating activity of FITC-fRunx2 compared with FITC-fRunx2 alone. In addition, the fRunx2-cLK group showed strong staining with alizarin red compared with other groups and the degree of alizarin red staining in the fRunx2-cLK group was also 1.2-fold higher than that in the fRunx2-Tat group. The ALP and osteocalcin gene expression levels in the fRunx2-cLK group were higher than those in the other groups. The fRunx2 transferred effectively into the cytoplasm aided by the cLK peptide and augmented the osteogenic differentiation of MSCs.
Collapse
|
25
|
Hydrogen Sulfide in Bone Tissue Regeneration and Repair: State of the Art and New Perspectives. Int J Mol Sci 2019; 20:ijms20205231. [PMID: 31652532 PMCID: PMC6834365 DOI: 10.3390/ijms20205231] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022] Open
Abstract
The importance of hydrogen sulfide (H2S) in the regulation of multiple physiological functions has been clearly recognized in the over 20 years since it was first identified as a novel gasotransmitter. In bone tissue H2S exerts a cytoprotective effect and promotes bone formation. Just recently, the scientific community has begun to appreciate its role as a therapeutic agent in bone pathologies. Pharmacological administration of H2S achieved encouraging results in preclinical studies in the treatment of systemic bone diseases, such as osteoporosis; however, a local delivery of H2S at sites of bone damage may provide additional opportunities of treatment. Here, we highlight how H2S stimulates multiple signaling pathways involved in various stages of the processes of bone repair. Moreover, we discuss how material science and chemistry have recently developed biomaterials and H2S-donors with improved features, laying the ground for the development of H2S-releasing devices for bone regenerative medicine. This review is intended to give a state-of-the-art description of the pro-regenerative properties of H2S, with a focus on bone tissue, and to discuss the potential of H2S-releasing scaffolds as a support for bone repair.
Collapse
|
26
|
Hydrogen sulfide-releasing silk fibroin scaffold for bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 102:471-482. [DOI: 10.1016/j.msec.2019.04.039] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 03/20/2019] [Accepted: 04/12/2019] [Indexed: 02/07/2023]
|
27
|
Iglesias-Echevarria M, Durante L, Johnson R, Rafuse M, Ding Y, Bonani W, Maniglio D, Tan W. Coaxial PCL/PEG-thiol-ene microfiber with tunable physico-chemical properties for regenerative scaffolds. Biomater Sci 2019; 7:3640-3651. [PMID: 31165794 PMCID: PMC6702085 DOI: 10.1039/c9bm00388f] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tissue regeneration requires scaffolds that exhibit mechanical properties similar to the tissues to be replaced while allowing cell infiltration and extracellular matrix production. Ideally, the scaffolds' porous architecture and physico-chemical properties can be precisely defined to address regenerative needs. We thus developed techniques to produce hybrid fibers coaxially structured with a polycaprolactone core and a 4-arm, polyethylene glycol thiol-norbornene sheath. We assessed the respective effects of crosslink density and sheath polymer size on the scaffold architecture, physical and mechanical properties, as well as cell-scaffold interactions in vitro and in vivo. All scaffolds displayed high elasticity, swelling and strength, mimicking soft tissue properties. Importantly, the thiol-ene hydrogel sheath enabled tunable softness and peptide tethering for cellular activities. With increased photopolymerization, stiffening and reduced swelling of scaffolds were found due to intra- and inter-fiber crosslinking. More polymerized scaffolds also enhanced the cell-scaffold interaction in vitro and induced spontaneous, deep cell infiltration to produce collagen and elastin for tissue regeneration in vivo. The molecular weight of sheath polymer provides an additional mechanism to alter the physical properties and biological activities of scaffolds. Overall, these robust scaffolds with tunable elasticity and regenerative cues offered a versatile and effective platform for tissue regeneration.
Collapse
|
28
|
Carvalho MS, Silva JC, Udangawa RN, Cabral JMS, Ferreira FC, da Silva CL, Linhardt RJ, Vashishth D. Co-culture cell-derived extracellular matrix loaded electrospun microfibrous scaffolds for bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 99:479-490. [PMID: 30889723 PMCID: PMC6452855 DOI: 10.1016/j.msec.2019.01.127] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 01/02/2023]
Abstract
Cell-derived extracellular matrix (ECM) has been employed as scaffolds for tissue engineering, creating a biomimetic microenvironment that provides physical, chemical and mechanical cues for cells and supports cell adhesion, proliferation, migration and differentiation by mimicking their in vivo microenvironment. Despite the enhanced bioactivity of cell-derived ECM, its application as a scaffold to regenerate hard tissues such as bone is still hampered by its insufficient mechanical properties. The combination of cell-derived ECM with synthetic biomaterials might result in an effective strategy to enhance scaffold mechanical properties and structural support. Electrospinning has been used in bone tissue engineering to fabricate fibrous and porous scaffolds, mimicking the hierarchical organized fibrillar structure and architecture found in the ECM. Although the structure of the scaffold might be similar to ECM architecture, most of these electrospun scaffolds have failed to achieve functionality due to a lack of bioactivity and osteoinductive factors. In this study, we developed bioactive cell-derived ECM electrospun polycaprolactone (PCL) scaffolds produced from ECM derived from human mesenchymal stem/stromal cells (MSC), human umbilical vein endothelial cells (HUVEC) and their combination based on the hypothesis that the cell-derived ECM incorporated into the PCL fibers would enhance the biofunctionality of the scaffold. The aims of this study were to fabricate and characterize cell-derived ECM electrospun PCL scaffolds and assess their ability to enhance osteogenic differentiation of MSCs, envisaging bone tissue engineering applications. Our findings demonstrate that all cell-derived ECM electrospun scaffolds promoted significant cell proliferation compared to PCL alone, while presenting similar physical/mechanical properties. Additionally, MSC:HUVEC-ECM electrospun scaffolds significantly enhanced osteogenic differentiation of MSCs as verified by increased ALP activity and osteogenic gene expression levels. To our knowledge, these results describe the first study suggesting that MSC:HUVEC-ECM might be developed as a biomimetic electrospun scaffold for bone tissue engineering applications.
Collapse
Affiliation(s)
- Marta S Carvalho
- Department of Bioengineering and iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal; Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA; The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal
| | - João C Silva
- Department of Bioengineering and iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal; Department of Chemistry and Chemical Biology, Biological Sciences and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA; The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal
| | - Ranodhi N Udangawa
- Department of Chemistry and Chemical Biology, Biological Sciences and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa 1049-001, Portugal
| | - Robert J Linhardt
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA; Department of Chemistry and Chemical Biology, Biological Sciences and Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA.
| | - Deepak Vashishth
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA.
| |
Collapse
|
29
|
Shih YV, Varghese S. Tissue engineered bone mimetics to study bone disorders ex vivo: Role of bioinspired materials. Biomaterials 2019; 198:107-121. [PMID: 29903640 PMCID: PMC6281816 DOI: 10.1016/j.biomaterials.2018.06.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/25/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022]
Abstract
Recent advances in materials development and tissue engineering has resulted in a substantial number of bioinspired materials that recapitulate cardinal features of bone extracellular matrix (ECM) such as dynamic inorganic and organic environment(s), hierarchical organization, and topographical features. Bone mimicking materials, as defined by its self-explanatory term, are developed based on the current understandings of the natural bone ECM during development, remodeling, and fracture repair. Compared to conventional plastic cultures, biomaterials that resemble some aspects of the native environment could elicit a more natural molecular and cellular response relevant to the bone tissue. Although current bioinspired materials are mainly developed to assist tissue repair or engineer bone tissues, such materials could nevertheless be applied to model various skeletal diseases in vitro. This review summarizes the use of bioinspired materials for bone tissue engineering, and their potential to model diseases of bone development and remodeling ex vivo. We largely focus on biomaterials, designed to re-create different aspects of the chemical and physical cues of native bone ECM. Employing these bone-inspired materials and tissue engineered bone surrogates to study bone diseases has tremendous potential and will provide a closer portrayal of disease progression and maintenance, both at the cellular and tissue level. We also briefly touch upon the application of patient-derived stem cells and introduce emerging technologies such as organ-on-chip in disease modeling. Faithful recapitulation of disease pathologies will not only offer novel insights into diseases, but also lead to enabling technologies for drug discovery and new approaches for cell-based therapies.
Collapse
Affiliation(s)
- Yuru Vernon Shih
- Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA.
| | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA; Department of Materials Science and Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
30
|
Zhu Y, Song F, Ju Y, Huang L, Zhang L, Tang C, Yang H, Huang C. NAC-loaded electrospun scaffolding system with dual compartments for the osteogenesis of rBMSCs in vitro. Int J Nanomedicine 2019; 14:787-798. [PMID: 30774333 PMCID: PMC6361317 DOI: 10.2147/ijn.s183233] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose In this study, we aimed to develop a unique N-acetyl cysteine (NAC)-loaded polylactic-co-glycolic acid (PLGA) electrospun system with separate compartments for the promotion of osteogenesis. Materials and methods We first prepared solutions of NAC-loaded mesoporous silica nanoparticles (MSNs), PLGA, and NAC in N, N-dimethylformamide and tetrahydrofuran for the construction of the electrospun system. We then fed solutions to a specific injector for electrospinning. The physical and chemical properties of the scaffold were characterized through scanning electron microscopy, transmission electron microscopy, and Fourier transform infrared spectroscopy. The release of NAC and Si from different PLGA scaffolds was estimated. The cell viability, cell growth, and osteogenic potential of rat bone marrow-derived stroma cell (rBMSCs) on different PLGA scaffolds were evaluated through MTT assay, live/dead staining, phalloidin staining, and Alizarin red staining. The expression levels of osteogenic-related markers were analyzed through real-time PCR (qRT-PCR). Results NAC was successfully loaded into MSNs. The addition of MSNs and NAC decreased the diameters of the electrospun fibers, increased the hydrophilicity and mechanical property of the PLGA scaffold. The release kinetic curve indicated that NAC was released from (PLGA + NAC)/(NAC@MSN) in a biphasic pattern, that featured an initial burst release stage and a later sustained release stage. This release pattern of NAC encapsulated on the (PLGA + NAC)/(NAC@MSN) scaffolds enabled to prolong the high concentrations of release of NAC, thus drastically affecting the osteogenic differentiation of rBMSCs. Conclusion A PLGA electrospun scaffold was developed, and MSNs were used as separate nanocarriers for recharging NAC concentration, demonstrating the promising use of (PLGA + NAC)/(NAC@MSN) for bone tissue engineering.
Collapse
Affiliation(s)
- Yuanjing Zhu
- Center for Smart Materials and Devices, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, Hubei, China,
| | - Fangfang Song
- Center for Smart Materials and Devices, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, Hubei, China,
| | - Yanyun Ju
- Center for Smart Materials and Devices, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, Hubei, China
| | - Liyuan Huang
- Center for Smart Materials and Devices, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, Hubei, China,
| | - Lu Zhang
- Center for Smart Materials and Devices, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, Hubei, China,
| | - Chuliang Tang
- Center for Smart Materials and Devices, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, Hubei, China,
| | - Hongye Yang
- Center for Smart Materials and Devices, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, Hubei, China,
| | - Cui Huang
- Center for Smart Materials and Devices, State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, School of Materials Science and Engineering, Wuhan University of Technology, Wuhan, Hubei, China,
| |
Collapse
|
31
|
Li YM, Wu JY, Jiang J, Dong SK, Chen YS, He HY, Liu CS, Zhao JZ. Chondroitin sulfate-polydopamine modified polyethylene terephthalate with extracellular matrix-mimetic immunoregulatory functions for osseointegration. J Mater Chem B 2019; 7:7756-7770. [DOI: 10.1039/c9tb01984g] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Optimal integration between the polyethylene terephthalate (PET) graft and host bone is a prerequisite to obtain a satisfactory outcome after graft implantation for ligament reconstruction.
Collapse
Affiliation(s)
- Ya-Min Li
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Jing-Yao Wu
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai
- China
| | - Jia Jiang
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Shi-Kui Dong
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Yun-Su Chen
- Department of Joint Surgery
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| | - Hong-Yan He
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai
- China
| | - Chang-Sheng Liu
- Engineering Research Center for Biomedical Materials of Ministry of Education
- East China University of Science and Technology
- Shanghai
- China
| | - Jin-Zhong Zhao
- Department of Sports Medicine
- Shanghai Jiaotong University Affiliated Sixth People's Hospital
- Shanghai 200233
- China
| |
Collapse
|
32
|
Wang X, Wang G, Zingales S, Zhao B. Biomaterials Enabled Cell-Free Strategies for Endogenous Bone Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2018; 24:463-481. [PMID: 29897021 DOI: 10.1089/ten.teb.2018.0012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Repairing bone defects poses a major orthopedic challenge because current treatments are constrained by the limited regenerative capacity of human bone tissue. Novel therapeutic strategies, such as stem cell therapy and tissue engineering, have the potential to enhance bone healing and regeneration, and hence may improve quality of life for millions of people. However, the ex vivo expansion of stem cells and their in vivo delivery pose technical difficulties that hamper clinical translation and commercial development. A promising alternative to cell delivery-based strategies is to stimulate or augment the inherent self-repair mechanisms of the patient to promote endogenous restoration of the lost/damaged bone. There is growing evidence indicating that increasing the endogenous regenerative potency of bone tissues for therapeutics will require the design and development of new generations of biomedical devices that provide key signaling molecules to instruct cell recruitment and manipulate cell fate for in situ tissue regeneration. Currently, a broad range of biomaterial-based deployment technologies are becoming available, which allow for controlled spatial presentation of biological cues required for endogenous bone regeneration. This article aims to explore the proposed concepts and biomaterial-enabled strategies involved in the design of cell-free endogenous techniques in bone regenerative medicine.
Collapse
Affiliation(s)
- Xiaojing Wang
- 1 Dental Implant Center, Affiliated Hospital of Qingdao University , Qingdao, P.R. China .,2 School of Stomatology, Qingdao University , Qingdao, Shandong, P.R. China
| | - Guowei Wang
- 3 Department of Stomatology, Laoshan Branch of No. 401 Hospital of the Chinese Navy , Qingdao, Shandong, P.R. China
| | - Sarah Zingales
- 4 Department of Chemistry and Biochemistry, Georgia Southern University , Savannah, Georgia
| | - Baodong Zhao
- 1 Dental Implant Center, Affiliated Hospital of Qingdao University , Qingdao, P.R. China .,2 School of Stomatology, Qingdao University , Qingdao, Shandong, P.R. China
| |
Collapse
|
33
|
Mohammadi M, Alibolandi M, Abnous K, Salmasi Z, Jaafari MR, Ramezani M. Fabrication of hybrid scaffold based on hydroxyapatite-biodegradable nanofibers incorporated with liposomal formulation of BMP-2 peptide for bone tissue engineering. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1987-1997. [PMID: 29933024 DOI: 10.1016/j.nano.2018.06.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/03/2018] [Accepted: 06/08/2018] [Indexed: 02/06/2023]
Abstract
In the present study, we fabricated an efficient, simple biomimetic scaffold to stimulate osteogenic differentiation of mesenchymal stem cells (MSCs). Electrospun poly L-lactic acid nanofibers were employed to mimic the nanofibrillar structure of bone proteins and coated with hydroxyapatite nanoparticles to simulate bone minerals. Thereafter, we regulated the release pattern of BMP-2 peptide through covalent attachment of an optimized liposomal formulation to the scaffold. The fabricated platform provided a sustained release profile of BMP-2 peptide up to 21 days while supporting cellular attachment and proliferation without cytotoxicity. In-vitro results confirmed the superiority of the scaffold containing liposomes through enhancement of growth and differentiation of MSCs. Ectopic bone formation model exhibited significant localized initiation of bone formation of liposome incorporated scaffold. Consequently, these findings demonstrated that our designed platform with modified release properties of BMP-2 peptide considerably promoted osteogenic differentiation of MSCs making it a unique candidate for bone regeneration therapeutics.
Collapse
Affiliation(s)
- Marzieh Mohammadi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical sciences, Mashhad, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
34
|
Buchaim DV, Bueno PCDS, Andreo JC, Roque DD, Roque JS, Zilio MG, Salatin JA, Kawano N, Furlanette G, Buchaim RL. Action of a deproteinized xenogenic biomaterial in the process of bone repair in rats submitted to inhalation of cigarette smoke. Acta Cir Bras 2018; 33:324-332. [PMID: 29768535 DOI: 10.1590/s0102-865020180040000004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/09/2018] [Indexed: 12/25/2022] Open
Abstract
PURPOSE To investigate if the inorganic bovine bone matrix changes the bone formation in rats submitted to inhalation of cigarette smoke. METHODS Twenty Wistar rats were divided into two groups: Cigarette Clot Group (CCG), which in the inhalation chamber received the smoke of 10 cigarettes, 3 times a day, 10 minutes, for 30 days and had the surgical cavity filled by clot; Cigarette Biomaterial Group (CBG), submitted to the same inhalation technique but with the cavity filled by biomaterial. RESULTS In CCG there was a significant difference of new bone tissue in the analyzed periods (15 and 45 days), and in 15 days, there was 4.8 ± 0.42 of bone formed and 11.73 ± 0.59 (p <0.05) in 45 days. The CBG also showed a significant difference between the periods of 15 to 45 days, being respectively 6.16 ± 0.30 and 11.60 ± 0.61. However, when the groups were compared, within the same analyzed periods, a significant difference was observed only in the period of 15 days, with the new bone percentage being greater in the CBG. CONCLUSION The bone matrix acted as an osteoinductive biomaterial, biocompatible and aided in the repair process, mainly in the initial period of recovery.
Collapse
Affiliation(s)
- Daniela Vieira Buchaim
- PhD, Medical and Dentistry School, Universidade de Marilia (UNIMAR), and Medical School, Centro Universitário de Adamantina (UNIFAI), Brazil. Conception and design of the study, manuscript preparation
| | | | - Jesus Carlos Andreo
- PhD, Department of Biological Sciences (Anatomy), Bauru School of Dentistry, Universidade de São Paulo (USP), Bauru-SP, Brazil. Acquisition, analysis and interpretation of data
| | - Domingos Donizeti Roque
- PhD, Medical School, UNIMAR, Marilia-SP, Brazil. Scientific and intellectual content of the study
| | - José Sidney Roque
- PhD, Department of Anatomy, Universidade Estadual do Norte do Paraná (UENP), Jacarezinho-PR, Brazil. Technical procedures
| | - Marcelo Garcia Zilio
- Graduate student, Medical School, UNIMAR, Marilia-SP, Brazil. Histopathological examinations, statistics analysis
| | - Jefferson Augusto Salatin
- Graduate student, Medical School, UNIMAR, Marilia-SP, Brazil. Histopathological examinations, statistics analysis
| | - Natália Kawano
- Graduate student, Medical School, UNIMAR, Marilia-SP, Brazil. Histopathological examinations, statistics analysis
| | - Gabriela Furlanette
- Graduate student, Medical School, UNIMAR, Marilia-SP, Brazil. Histopathological examinations, statistics analysis
| | - Rogério Leone Buchaim
- PhD, Medical and Dentistry School, UNIMAR, Marilia-SP, and USP, Bauru-SP, Brazil. Manuscript preparation and writing, critical revision, final approval
| |
Collapse
|
35
|
Ding Y, Li W, Correia A, Yang Y, Zheng K, Liu D, Schubert DW, Boccaccini AR, Santos HA, Roether JA. Electrospun Polyhydroxybutyrate/Poly(ε-caprolactone)/Sol-Gel-Derived Silica Hybrid Scaffolds with Drug Releasing Function for Bone Tissue Engineering Applications. ACS APPLIED MATERIALS & INTERFACES 2018; 10:14540-14548. [PMID: 29624366 PMCID: PMC6108537 DOI: 10.1021/acsami.8b02656] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/06/2018] [Indexed: 05/23/2023]
Abstract
Electrospun hybrid scaffolds are an effective platform to deliver drugs site specifically for the prevention and treatment of diseases in addition to promote tissue regeneration because of the flexibility to load drugs therein. In the present study, electrospun hybrid scaffolds containing antibiotics were developed to support cellular activities and eliminate potential postoperative inflammation and infection. As a model drug, levofloxacin (LFX) was successfully incorporated into pure polyhydroxybutyrate/poly(ε-caprolactone) (PHB/PCL) scaffolds and PHB/PCL/sol-gel-derived silica (SGS) scaffolds. The influence of LFX on the morphology, mechanical performance, chemical structure, drug release profile, and antibacterial effect of the scaffolds was thoroughly and comparatively investigated. MG-63 osteoblast-like cell cultivation on both scaffolds certified that LFX inclusion did not impair the biocompatibility. In addition to the favorable cellular proliferation and differentiation, scaffolds containing both LFX and SGS displayed highly increased mineralization content. Therefore, the present multifunctional hybrid scaffolds are promising in tissue engineering applications.
Collapse
Affiliation(s)
- Yaping Ding
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
- Institute of Polymer Materials, University of Erlangen−Nuremberg, Martensstrasse 7, 91058 Erlangen, Germany
| | - Wei Li
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| | - Alexandra Correia
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| | - Yuyun Yang
- Institute of Biomaterials, University of
Erlangen−Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
- Institute of Corrosion
Science and Surface Technology, Harbin Engineering
University, Nantong Street 145, 150001 Harbin, China
| | - Kai Zheng
- Institute of Biomaterials, University of
Erlangen−Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
| | - Dongfei Liu
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| | - Dirk W. Schubert
- Institute of Polymer Materials, University of Erlangen−Nuremberg, Martensstrasse 7, 91058 Erlangen, Germany
| | - Aldo R. Boccaccini
- Institute of Biomaterials, University of
Erlangen−Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany
| | - Hélder A. Santos
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, and Helsinki Institute of Life Science (HiLIFE), University of Helsinki, FI-00014 Helsinki, Finland
| | - Judith A. Roether
- Institute of Polymer Materials, University of Erlangen−Nuremberg, Martensstrasse 7, 91058 Erlangen, Germany
| |
Collapse
|
36
|
Lu L, Liu B, Shang X, Zhang Y, Chen W, Liu S, Huang J, Wang A, Guo Q, Lu S. [Construction of tissue engineered cartilage based on acellular cartilage extracellular matrix oriented scaffold and chondrocytes]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:291-297. [PMID: 29806277 DOI: 10.7507/1002-1892.201710095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Objective To observe the feasibility of acellular cartilage extracellular matrix (ACECM) oriented scaffold combined with chondrocytes to construct tissue engineered cartilage. Methods Chondrocytes from the healthy articular cartilage tissue of pig were isolated, cultured, and passaged. The 3rd passage chondrocytes were labeled by PKH26. After MTT demonstrated that PKH26 had no influence on the biological activity of chondrocytes, labeled and unlabeled chondrocytes were seeded on ACECM oriented scaffold and cultivated. The adhesion, growth, and distribution were evaluated by gross observation, inverted microscope, and fluorescence microscope. Scanning electron microscope was used to observe the cellular morphology after cultivation for 3 days. Type Ⅱ collagen immunofluorescent staining was used to check the secretion of extracellular matrix. In addition, the complex of labeled chondrocytes and ACECM oriented scaffold (cell-scaffold complex) was transplanted into the subcutaneous tissue of nude mouse. After transplantation, general physical conditions of nude mouse were observed, and the growth of cell-scaffold complex was observed by molecular fluorescent living imaging system. After 4 weeks, the neotissue was harvested to analyze the properties of articular cartilage tissue by gross morphology and histological staining (Safranin O staining, toluidine blue staining, and typeⅡcollagen immunohistochemical staining). Results After chondrocytes that were mainly polygon and cobblestone like shape were seeded and cultured on ACECM oriented scaffold for 7 days, the neotissue was translucency and tenacious and cells grew along the oriented scaffold well by inverted microscope and fluorescence microscope. In the subcutaneous microenvironment, the cell-scaffold complex was cartilage-like tissue and abundant cartilage extracellular matrix (typeⅡcollagen) was observed by histological staining and typeⅡcollagen immunohistochemical staining. Conclusion ACECM oriented scaffold is benefit to the cell adhesion, proliferation, and oriented growth and successfully constructes the tissue engineered cartilage in nude mouse model, which demonstrates that the ACECM oriented scaffold is promise to be applied in cartilage tissue engineering.
Collapse
Affiliation(s)
- Liang Lu
- Department of Orthopedics, Anhui Provincial Hospital, Hefei Anhui, 230001, P.R.China
| | - Bin Liu
- Department of Orthopedics, Anhui Provincial Hospital, Hefei Anhui, 230001, P.R.China
| | - Xifu Shang
- Department of Orthopedics, Anhui Provincial Hospital, Hefei Anhui, 230001, P.R.China
| | - Yu Zhang
- Department of Orthopedics, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing Jiangsu, 210008, P.R.China;Orthopedics Research Institute, General Hospital of Chinese PLA, Beijing, 100853, P.R.China
| | - Weijian Chen
- Department of Orthopedics, Anhui Provincial Hospital, Hefei Anhui, 230001, P.R.China
| | - Shuyun Liu
- Orthopedics Research Institute, General Hospital of Chinese PLA, Beijing, 100853, P.R.China
| | - Jingxiang Huang
- Orthopedics Research Institute, General Hospital of Chinese PLA, Beijing, 100853, P.R.China
| | - Aiyuan Wang
- Orthopedics Research Institute, General Hospital of Chinese PLA, Beijing, 100853, P.R.China
| | - Quanyi Guo
- Orthopedics Research Institute, General Hospital of Chinese PLA, Beijing, 100853,
| | - Shibi Lu
- Orthopedics Research Institute, General Hospital of Chinese PLA, Beijing, 100853, P.R.China
| |
Collapse
|
37
|
Mohammadi M, Mousavi Shaegh SA, Alibolandi M, Ebrahimzadeh MH, Tamayol A, Jaafari MR, Ramezani M. Micro and nanotechnologies for bone regeneration: Recent advances and emerging designs. J Control Release 2018; 274:35-55. [PMID: 29410062 DOI: 10.1016/j.jconrel.2018.01.032] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 02/08/2023]
Abstract
Treatment of critical-size bone defects is a major medical challenge since neither the bone tissue can regenerate nor current regenerative approaches are effective. Emerging progresses in the field of nanotechnology have resulted in the development of new materials, scaffolds and drug delivery strategies to improve or restore the damaged tissues. The current article reviews promising nanomaterials and emerging micro/nano fabrication techniques for targeted delivery of biomolecules for bone tissue regeneration. In addition, recent advances in fabrication of bone graft substitutes with similar properties to normal tissue along with a brief summary of current commercialized bone grafts have been discussed.
Collapse
Affiliation(s)
- Marzieh Mohammadi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Ali Mousavi Shaegh
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Clinical Research Unit, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ali Tamayol
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE 68588, USA; Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02139, USA
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
38
|
Sphingosine 1-phosphate (S1P) signalling: Role in bone biology and potential therapeutic target for bone repair. Pharmacol Res 2017; 125:232-245. [PMID: 28855094 DOI: 10.1016/j.phrs.2017.08.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/30/2022]
Abstract
The lipid mediator sphingosine 1-phosphate (S1P) affects cellular functions in most systems. Interest in its therapeutic potential has increased following the discovery of its G protein-coupled receptors and the recent availability of agents that can be safely administered in humans. Although the role of S1P in bone biology has been the focus of much less research than its role in the nervous, cardiovascular and immune systems, it is becoming clear that this lipid influences many of the functions, pathways and cell types that play a key role in bone maintenance and repair. Indeed, S1P is implicated in many osteogenesis-related processes including stem cell recruitment and subsequent differentiation, differentiation and survival of osteoblasts, and coupling of the latter cell type with osteoclasts. In addition, S1P's role in promoting angiogenesis is well-established. The pleiotropic effects of S1P on bone and blood vessels have significant potential therapeutic implications, as current therapeutic approaches for critical bone defects show significant limitations. Because of the complex effects of S1P on bone, the pharmacology of S1P-like agents and their physico-chemical properties, it is likely that therapeutic delivery of S1P agents will offer significant advantages compared to larger molecular weight factors. Hence, it is important to explore novel methods of utilizing S1P agents therapeutically, and improve our understanding of how S1P and its receptors modulate bone physiology and repair.
Collapse
|
39
|
Current approaches for modulation of the nanoscale interface in the regulation of cell behavior. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 14:2455-2464. [PMID: 28552647 PMCID: PMC6173683 DOI: 10.1016/j.nano.2017.03.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/20/2017] [Accepted: 03/09/2017] [Indexed: 11/22/2022]
Abstract
Regulation of cell behavior in response to nanoscale features has been the focus of much research in recent years and the successful generation of nanoscale features capable of mimicking the natural nanoscale interface has been of great interest in the field of biomaterials research. In this review, we discuss relevant nanofabrication techniques and how they are combined with bioengineering applications to mimic the natural extracellular matrix (ECM) and create valuable nanoscale interfaces.
Collapse
|
40
|
Xing J, Mei T, Luo K, Li Z, Yang A, Li Z, Xie Z, Zhang Z, Dong S, Hou T, Xu J, Luo F. A nano-scaled and multi-layered recombinant fibronectin/cadherin chimera composite selectively concentrates osteogenesis-related cells and factors to aid bone repair. Acta Biomater 2017; 53:470-482. [PMID: 28193541 DOI: 10.1016/j.actbio.2017.02.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 02/05/2017] [Accepted: 02/09/2017] [Indexed: 01/06/2023]
Abstract
Easily accessible and effective bone grafts are in urgent need in clinic. The selective cell retention (SCR) strategy, by which osteogenesis-related cells and factors are enriched from bone marrow into bio-scaffolds, holds great promise. However, the retention efficacy is limited by the relatively low densities of osteogenesis-related cells and factors in marrow; in addition, a lack of satisfactory surface modifiers for scaffolds further exacerbates the dilemma. To address this issue, a multi-layered construct consisting of a recombinant fibronectin/cadherin chimera was established via a layer-by-layer self-assembly technique (LBL-rFN/CDH) and used to modify demineralised bone matrix (DBM) scaffolds. The modification was proven stable and effective. By the mechanisms of physical interception and more importantly, chemical recognition (fibronectin/integrins), the LBL-rFN/CDH modification significantly improved the retention efficacy and selectivity for osteogenesis-related cells, e.g., monocytes, mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs), and bioactive factors, e.g., bFGF, BMP-2 and SDF-1α. Moreover, the resulting composite (designated as DBM-LBL-rFN/CDH) not only exhibited a strong MSC-recruiting capacity after SCR, but also provided favourable microenvironments for the proliferation and osteogenic differentiation of MSCs. Eventually, bone repair was evidently improved. Collectively, DBM-LBL-rFN/CDH presented a suitable biomaterial for SCR and a promising solution for tremendous need for bone grafts. STATEMENT OF SIGNIFICANCE There is an urgent need for effective bone grafts. With the potential of integrating osteogenicity, osteoinductivity and osteoconductivity, selective cell retention (SCR) technology brings hope for developing ideal grafts. However, it is constrained by low efficacy and selectivity. Thus, we modified demineralized bone matrix with nano-scaled and multi-layered recombinant fibronectin/cadherin chimera (DBM-rFN/CDH-LBL), and evaluate its effects on SCR and bone repair. DBM-rFN/CDH-LBL significantly improved the efficacy and selectivity of SCR via physical interception and chemical recognition. The post-enriched DBM-rFN/CDH-LBL provided favourable microenvironments to facilitate the migration, proliferation and osteogenic differentiation of MSCs, thus accelerating bone repair. Conclusively, DBM-rFN/CDH-LBL presents a novel biomaterial with advantages including high cost-effectiveness, more convenience for storage and transport and can be rapidly constructed intraoperatively.
Collapse
Affiliation(s)
- Junchao Xing
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Tieniu Mei
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Keyu Luo
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Zhiqiang Li
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Aijun Yang
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Zhilin Li
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China; Department of Spine, Lanzhou General Hospital, Lanzhou Command of CPLA, Lanzhou 730050, China
| | - Zhao Xie
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Zehua Zhang
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Shiwu Dong
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China; Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Tianyong Hou
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China
| | - Jianzhong Xu
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China.
| | - Fei Luo
- Department of Orthopedics, National & Regional United Engineering Laboratory of Tissue Engineering, Southwest Hospital, the Third Military Medical University, Chongqing, China; Center of Regenerative and Reconstructive Engineering Technology in Chongqing City, Chongqing, China; Tissue Engineering Laboratory of Chongqing City, Chongqing, China.
| |
Collapse
|
41
|
Lagonegro P, Rossi F, Galli C, Smerieri A, Alinovi R, Pinelli S, Rimoldi T, Attolini G, Macaluso G, Macaluso C, Saddow S, Salviati G. A cytotoxicity study of silicon oxycarbide nanowires as cell scaffold for biomedical applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 73:465-471. [DOI: 10.1016/j.msec.2016.12.096] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/16/2016] [Accepted: 12/20/2016] [Indexed: 12/20/2022]
|
42
|
Mechiche Alami S, Rammal H, Boulagnon-Rombi C, Velard F, Lazar F, Drevet R, Laurent Maquin D, Gangloff S, Hemmerlé J, Voegel J, Francius G, Schaaf P, Boulmedais F, Kerdjoudj H. Harnessing Wharton's jelly stem cell differentiation into bone-like nodule on calcium phosphate substrate without osteoinductive factors. Acta Biomater 2017; 49:575-589. [PMID: 27888100 DOI: 10.1016/j.actbio.2016.11.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/11/2016] [Accepted: 11/17/2016] [Indexed: 01/05/2023]
Abstract
An important aim of bone regenerative medicine is to design biomaterials with controlled chemical and topographical features to guide stem cell fate towards osteoblasts without addition of specific osteogenic factors. Herein, we find that sprayed bioactive and biocompatible calcium phosphate substrates (CaP) with controlled topography induce, in a well-orchestrated manner, Wharton's jelly stem cells (WJ-SCs) differentiation into osteoblastic lineage without any osteogenic supplements. The resulting WJ-SCs commitment exhibits features of native bone, through the formation of three-dimensional bone-like nodule with osteocyte-like cells embedded into a mineralized type I collagen. To our knowledge, these results present the first observation of a whole differentiation process from stem cell to osteocytes-like on a synthetic material. This suggests a great potential of sprayed CaP and WJ-SCs in bone tissue engineering. These unique features may facilitate the transition from bench to bedside and the development of successful engineered bone. STATEMENT OF SIGNIFICANCE Designing materials to direct stem cell fate has a relevant impact on stem cell biology and provides insights facilitating their clinical application in regenerative medicine. Inspired by natural bone compositions, a friendly automated spray-assisted system was used to build calcium phosphate substrate (CaP). Sprayed biomimetic solutions using mild conditions led to the formation of CaP with controlled physical properties, good bioactivity and biocompatibility. Herein, we show that via optimization of physical properties, CaP substrate induce osteogenic differentiation of Wharton's jelly stem cells (WJ-SCs) without adding osteogenic supplement factors. These results suggest a great potential of sprayed CaP and WJ-SCs in bone tissue engineering and may facilitate the transition from bench to beside and the development of clinically successful engineered bone.
Collapse
|
43
|
Blair HC, Larrouture QC, Li Y, Lin H, Beer-Stoltz D, Liu L, Tuan RS, Robinson LJ, Schlesinger PH, Nelson DJ. Osteoblast Differentiation and Bone Matrix Formation In Vivo and In Vitro. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:268-280. [PMID: 27846781 DOI: 10.1089/ten.teb.2016.0454] [Citation(s) in RCA: 288] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We review the characteristics of osteoblast differentiation and bone matrix synthesis. Bone in air breathing vertebrates is a specialized tissue that developmentally replaces simpler solid tissues, usually cartilage. Bone is a living organ bounded by a layer of osteoblasts that, because of transport and compartmentalization requirements, produce bone matrix exclusively as an organized tight epithelium. With matrix growth, osteoblasts are reorganized and incorporated into the matrix as living cells, osteocytes, which communicate with each other and surface epithelium by cell processes within canaliculi in the matrix. The osteoblasts secrete the organic matrix, which are dense collagen layers that alternate parallel and orthogonal to the axis of stress loading. Into this matrix is deposited extremely dense hydroxyapatite-based mineral driven by both active and passive transport and pH control. As the matrix matures, hydroxyapatite microcrystals are organized into a sophisticated composite in the collagen layer by nucleation in the protein lattice. Recent studies on differentiating osteoblast precursors revealed a sophisticated proton export network driving mineralization, a gene expression program organized with the compartmentalization of the osteoblast epithelium that produces the mature bone matrix composite, despite varying serum calcium and phosphate. Key issues not well defined include how new osteoblasts are incorporated in the epithelial layer, replacing those incorporated in the accumulating matrix. Development of bone in vitro is the subject of numerous projects using various matrices and mesenchymal stem cell-derived preparations in bioreactors. These preparations reflect the structure of bone to variable extents, and include cells at many different stages of differentiation. Major challenges are production of bone matrix approaching the in vivo density and support for trabecular bone formation. In vitro differentiation is limited by the organization and density of osteoblasts and by endogenous and exogenous inhibitors.
Collapse
Affiliation(s)
- Harry C Blair
- 1 Veteran's Affairs Medical Center , Pittsburgh, Pennsylvania.,2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | | | - Yanan Li
- 3 Department of Stomatology, Chinese PLA General Hospital , Beijing, China
| | - Hang Lin
- 4 Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Donna Beer-Stoltz
- 2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Li Liu
- 2 Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Rocky S Tuan
- 4 Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Lisa J Robinson
- 5 Department of Pathology, West Virginia University School of Medicine , Morgantown, West Virginia.,6 Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Paul H Schlesinger
- 7 Department of Cell Biology, Washington University , Saint Louis, Missouri
| | - Deborah J Nelson
- 8 Department of Neurobiology, Pharmacology & Physiology, University of Chicago , Chicago, Illinois
| |
Collapse
|